Vous êtes sur la page 1sur 10

feature articles

Reviews and

Molecular mechanisms in allergy and clinical immunology


Series editors: Joshua A. Boyce, MD, Fred Finkelman, MD, William T. Shearer, MD, PhD, and Donata
Vercelli, MD

Mechanisms of allergen-specific
immunotherapy
Mübeccel Akdis, MD, PhD, and Cezmi A. Akdis, MD Davos, Switzerland

This activity is available for CME credit. See page 48A for important information.

Allergen-specific immunotherapy (SIT) has been used for circulating basophils takes place. Current understanding of
almost a century as a desensitizing therapy for allergic diseases mechanisms of allergen-SIT, particularly the role of Treg cells
and represents the only curative and specific method of in peripheral tolerance, may enable novel treatment strategies.
treatment. Administration of appropriate concentrations of (J Allergy Clin Immunol 2007;119:780-9.)
allergen extracts has been shown to be reproducibly effective
when patients are carefully selected. The mechanisms by which Key words: Allergen immunotherapy, T-regulatory cells, tolerance,
allergen-SIT has its effects include the modulation of T-cell and IgE, IgG, T cells, B cells, mast cells, basophils, eosinophils, IL-10,
B-cell responses and related antibody isotypes as well as TGF-b
effector cells of allergic inflammation, such as eosinophils,
basophils, and mast cells. The balance between allergen-specific
T-regulatory (Treg) and TH2 cells appears to be decisive in the The physiopathology of allergic immune responses is
development of allergic and healthy immune responses against complex and has been shown to be influenced by several
allergens. Treg cells consistently represent the dominant subset
factors, including genetic susceptibility, route of expo-
specific for common environmental allergens in sensitized
healthy individuals. In contrast, there is a high frequency of sure, allergen dose, and, in some cases, the structural
allergen-specific TH2 cells in patients with allergy. The characteristics of the allergen.1 Allergic immune response
induction of a tolerant state in peripheral T cells represents an requires sensitization and development of specific im-
essential step in allergen-SIT. Peripheral T-cell tolerance is mune response toward the allergen. During sensitization
characterized mainly by generation of allergen-specific Treg to allergen, priming of allergen-specific CD41 TH2 cells
cells leading to suppressed T-cell proliferation and TH1 and results in the production of TH2 cytokines (such as IL-4
TH2 cytokine responses against the allergen. This is and IL-13) that are responsible for class switching to
accompanied by a significant increase in allergen-specific IgG4, the e heavy chain for IgE production by B cells, mucus
and also IgG1 and IgA, and a decrease in IgE in the late stage of production, and activation of endothelial cells for TH2
the disease. In addition, decreased tissue infiltration of mast
cell and eosinophil migration to tissues.2,3 IgE sensitizes
cells and eosinophils and their mediator release including
mast cells and basophils by binding to the high-affinity
receptor for IgE (FceRI) expressed on their surface. On
cross-linking of the IgE-FceRI complexes by allergen,
From the Swiss Institute of Allergy and Asthma Research. mast cells and basophils degranulate, releasing vasoactive
The authors’ laboratories are supported by Swiss National Science Foundation
amines (principally histamine), lipid mediators (prosta-
grants 32-112306 and 32-105268 and the Global Allergy and Asthma
European Network. glandins and cysteinyl leukotrienes), cytokines, and che-
Disclosure of potential conflict of interest: M. Akdis has received grant support mokines, all of which characterize the immediate phase
from ALK-Abelló, Stallergenes, Allergopharma Joachim Ganzer KG, Essex of the allergic reaction.2,3 After the sensitization phase,
Chemie, Allecure, the Global Allergy Asthma European Network, and the allergic inflammation and reactions to allergen challenge
Swiss National Science Foundation and is on the speakers’ bureau for
ALK-Abelló. C. A. Akdis has received grant support from ALK-Abelló,
are observed in the target organ, leading to development
Stallergenes, Allergopharma Joachim Ganzer KG, Essex Chemie, of allergic rhinoconjunctivitis, eczema, asthma, or sys-
Allecure, the Global Allergy Asthma European Network, and the Swiss temic anaphylaxis. T cells constitute a large population
National Science Foundation and is on the speakers’ bureau for Stallergenes, of cellular infiltrate in allergic inflammation, and a dys-
Allergopharma Joachim Ganzer KG, and Merck.
regulated immune response appears to be an important
Received for publication December 11, 2006; revised January 14, 2007;
accepted for publication January 16, 2007. pathogenetic factor. Cardinal events during allergic in-
Available online March 6, 2007. flammation can be classified as activation, organ-selective
Reprint requests: Cezmi A. Akdis, MD, Swiss Institute of Allergy and Asthma homing, survival and reactivation, and effector functions
Research, Obere Strasse 22, CH7270 Davos, Switzerland. E-mail: akdisac@ of immune system cells.4 T cells are activated by aeroaller-
siaf.unizh.ch.
0091-6749/$32.00
gens, food antigens, autoantigens, and bacterial superanti-
Ó 2007 American Academy of Allergy, Asthma & Immunology gens in allergic inflammation. They are under the influence
doi:10.1016/j.jaci.2007.01.022 of skin, lung, or nose-related chemokine networks, and
780
J ALLERGY CLIN IMMUNOL Akdis and Akdis 781
VOLUME 119, NUMBER 4

feature articles
Reviews and
TABLE I. Effects of allergen-SIT on clinical and
Abbreviations used immunologic parameters
LPR: Late-phase reaction
Clinical Long-term cure
PIT: Peptide immunotherapy
parameters Decreased clinical symptoms and drug use
SIT: Specific immunotherapy
Decreased response to allergen challenge tests
SLIT: Sublingual immunotherapy
Decreased size and cellular influx in skin LPR
TLR: Toll-like receptor
Decreased skin type I hypersensitivity response
Tr1: Type 1 T-regulatory
Improved quality of life
Treg: T-regulatory
Mast cells Very early desensitization effect
Reduction of tissue numbers
Decrease in mediator release
Decrease in proinflammatory cytokine production
Basophils Very early desensitization effect
they show organ-selective homing. A prolonged survival Decrease in mediator release
of the inflammatory cells in the tissues and consequent Decrease in proinflammatory cytokine production
reactivation is observed in the subepithelial tissues. Finally, Eosinophils Reduction of tissue numbers
T cells display effector functions, which result in the Decrease in mediator release
induction of hyper-IgE, eosinophil survival, and mucus T cells Decreased allergen-induced proliferation
Induction of Treg cells
hyperproduction; and interact with bronchial epithelial
Increased secretion of IL-10 and TGF-b
cells, smooth muscle cells, and keratinocytes, causing Suppression of TH2 cells and cytokines
their activation and apoptosis.4,5 Peripheral T-cell toler- Decreased T-cell numbers in LPR
ance to allergens can overcome all of these pathological B cells Early increase and late decrease in serum
events in allergic inflammation because they all require specific IgE
T-cell activation. Increased serum specific IgG4
Increased serum specific IgG1 (relatively low
compared with IgG4)
SEQUENTIAL EVENTS IN ALLERGEN- Increased serum specific IgA
SPECIFIC IMMUNOTHERAPY AND THEIR Suppressed IgE-facilitated antigen presentation
UNDERLYING MECHANISMS Dendritic cells Different subsets can be targeted depending on
the type of the adjuvant
Very early desensitization effect Suppressed IgE-facilitated antigen presentation
The underlying immunological mechanisms of aller- Monocytes Increased IL-10 production
gen-specific immunotherapy (SIT) are continuously being
elucidated (Table I; Fig 1). Very early effects are related
to mast cell and basophil desensitization. Intermediate during the course of allergen-SIT, the suppression of mast
effects are related to changes in allergen-specific T cells, cells and basophils continues to be affected by changes
and late affects are related to B cells and IgE as well as in other immune parameters such as the generation of
mast cells, basophils, and eosinophils. Although definite allergen-specific T-regulatory (Treg) cells and decreased
decrease in IgE antibody levels and IgE-mediated skin specific IgE.
sensitivity normally requires years of SIT, most patients
are protected against bee stings already at an early stage Generation of Treg cells and peripheral
of venom-SIT. An important observation starting from T-cell tolerance
the first injection is an early decrease in mast cell and ba- The induction of a tolerant state in peripheral T cells
sophil activity for degranulation and systemic anaphylaxis represents an essential step in allergen-SIT (Table I; Fig
(Fig 1). Although it seems similar to rapid desensitization 1). Peripheral T-cell tolerance is characterized mainly
for hypersensitivity reactions to drugs, the mechanism by generation of allergen-specific Treg cells, suppressed
of this desensitization effect is yet unknown. Acute oral proliferative and cytokine responses against the major
desensitization to penicillin V in mice demonstrated that an- allergen (Fig 2).10-12 Subsets of Treg cells with distinct
tigen-specific mast cell desensitization is one of the main un- phenotypes and mechanisms of action include the natu-
derlying mechanisms for oral desensitization.6 It has been rally occurring thymic selected CD41CD251 Treg cells
shown that mediators of anaphylaxis (histamine and leuko- and the inducible type 1 Treg (Tr1) cells.13 In allergen-
trienes) are released during SIT and sting challenges without SIT, peripheral T-cell tolerance is initiated by autocrine
inducing a systemic anaphylactic response.7 Their piece- action of IL-10 and TGF-b, which is increasingly pro-
meal release below the threshold of systemic anaphylaxis duced by the antigen-specific T cells.11,14,15 The suppres-
may decrease the granule content of mediators and also sion by these cells could be partially blocked by the use
may affect the threshold of activation of mast cells and ba- of neutralizing antibodies against secreted or membrane-
sophils, because decreased mediator release in these cells bound IL-10 and TGF-b.12 However, these cells do ex-
is a well demonstrated feature in in vitro analysis a short press CD4 and CD25, raising the question whether these
time after the start of allergen-SIT.7-9 Although there are are inducible Tr1 cells, which have upregulated CD25,
fluctuations and a risk for developing systemic anaphylaxis or naturally occurring CD41CD251 Treg cells that
782 Akdis and Akdis J ALLERGY CLIN IMMUNOL
APRIL 2007
feature articles
Reviews and

FIG 1. Immunologic changes during the course of allergen-SIT. Although there is significant variation between
donors and protocols, starting from the first injection, an early decrease in mast cell and basophil activity and
degranulation and decreased tendency for systemic anaphylaxis is observed. This is followed by generation
of allergen-specific Treg cells and suppression of allergen-specific TH1 and TH2 cells. An early increase of spe-
cific IgE and late decrease is observed. This is in parallel to an increase of IgG4 in particular and, in some stud-
ies, IgG1 and IgA. A significant decrease in the allergen-specific IgE/IgG4 ratio occurs after several months. A
significant decrease in type I skin test reactivity is also observed relatively late in the course. Decrease in tissue
mast cells and eosinophils and release of their mediators and decrease in LPR is observed after a few months.

produce suppressive cytokines.13 In addition, it has been lead to either allergy development or recovery. Another
shown that CD41CD251 Treg cells from atopic donors study on healthy immune response to allergens demon-
have a reduced capability to suppress the proliferation of strated that CD41CD251 Treg cells have been associated
CD41CD252 T cells.16 Therefore, it has been suggested with the spontaneous remission of cow’s milk allergy.
that upregulation of CD41CD251 Treg cells plays a role Children who outgrew their allergy (tolerant children)
in allergen-SIT. TGF-b plays a dual role in allergic dis- had higher frequencies of circulating CD41CD251 T cells
ease: it suppresses allergen-specific T cells and plays a and decreased in vitro proliferative responses to bovine
role in remodeling of the tissues. It remains to be eluci- b-lactoglobulin in PBMCs compared with children who
dated in allergic inflammation whether the remodeling maintained clinically active allergy.20 Several studies
and the suppressive roles of TGF-b show an imbalance have been reported in other diseases in the same line.
that aggravates the disease instead of controlling the The in vitro proliferative response to nickel of human
immune response.17 CD41 T cells from healthy, nonallergic individuals was
Studies on the mechanisms by which immune re- strongly augmented when CD41CD251 Treg cells were
sponses to nonpathogenic environmental antigens lead depleted.21 Furthermore, a human in vivo study on immu-
to either allergy or nonharmful immunity have demon- notherapy of rheumatoid arthritis also showed a marked
strated that Treg cells are dominant in healthy individ- increase in the number of FoxP31CD41CD251 Treg cells
uals.18,19 If a detectable immune response is mounted, Tr1 in peripheral blood.22 CD251 Treg cells are characterized
cells specific for common environmental allergens consis- by the expression of the transcriptional regulator Foxp3
tently represent the dominant subset in healthy individ- (FOXP3 in human beings), which appears to be the master
uals. They use multiple suppressive mechanisms, IL-10 switch gene for Treg cell development and function. The
and TGF-b as secreted cytokines, and cytotoxic T-lym- spontaneous development of allergic airway inflamma-
phocyte antigen 4 and programmed death 1 as surface tion, hyper-IgE, and eosinophilia in addition to various
molecules. Healthy and allergic individuals exhibit all autoimmune diseases in Foxp3 mutant mice provides
3—TH1, TH2, and Tr1 allergen-specific subsets—in dif- compelling evidence for its importance in allergic inflam-
ferent proportions.18 Accordingly, a change in the dominant mation.23 Human beings with immune dysregulation
subset and the balance between TH2 and Treg cells may polyendocrinopathy enteropathy X-linked syndrome are
J ALLERGY CLIN IMMUNOL Akdis and Akdis 783
VOLUME 119, NUMBER 4

feature articles
Reviews and

FIG 2. A, Suppression of TH2 cell-mediated features of allergic inflammation by Treg cells. Treg cells use mul-
tiple suppressor factors to regulate undesired activity of effector TH2 cells. IL-10 and TGF-b suppress IgE pro-
duction and induce IgG4 and IgA, respectively. Both cytokines directly suppress allergic inflammation induced
by effector cells such as mast cells, basophils, and eosinophils. In addition, TH2 cells are suppressed by Treg
cells and can therefore no longer provide cytokines such as IL-3, IL-4, IL-5, IL-9, and IL-13. These cytokines are
required for the differentiation, survival, and activity of mast cells, basophils, eosinophils, and mucus-produc-
ing cells, as well as for the tissue homing of TH2 cells and eosinophils (red line, suppression; black line, stim-
ulation). B, Suppression of TH1 cell-mediated features of allergic inflammation by Treg cells. The TH1 cytokine,
IFN-g, in combination with TNF-a and/or FasL, induces apoptosis of smooth muscle cells, keratinocytes, and
bronchial epithelial cells as essential tissue injury events in atopic dermatitis and asthma (red line, suppres-
sion; black line, stimulation). FasL, Fas-ligand; IP-10, IFN-g–inducible protein 10; mig, monokine induced by
IFN-g; iTac, IFN-g–inducible chemoattractant.
784 Akdis and Akdis J ALLERGY CLIN IMMUNOL
APRIL 2007
feature articles
Reviews and

similarly affected and mostly develop hyper-IgE and Recently, a novel assay that detects allergen-IgE binding
eczema because of mutations in the FOXP3 gene.23 Sup- by using flow cytometry has been used to detect functional
porting these findings, a dysregulation of disease-causing SIT-induced changes in IgG antibody activity. Results
effector T cells is observed in atopic dermatitis lesions in suggest that successful SIT is associated with an increase
association with an impaired CD41CD251FoxP31 T-cell in IgG blocking activity that is not solely dependent on the
infiltration in the dermis.24 quantity of IgG antibodies.32,33 It seems to be relevant to
The role of Treg cells is not limited to suppression of measure the blocking activity of allergen-specific IgG or
TH2 cells. Peripheral tolerance uses multiple mechanisms IgG subsets, particularly IgG4 and also IgG1, instead of
to suppress allergic inflammation. Apparently, Treg cells the crude levels in sera. In this context, the role of anti-
contribute to the control of allergen-specific immune re- IgE treatment in the induction phase of allergen-SIT on
sponses in 5 major ways: suppression of antigen-present- safety and efficacy has been questioned. Anti-IgE mAb
ing cells that support the generation of effector TH2 and pretreatment enhances the safety of SIT for allergic rhinitis
TH1 cells; suppression of TH2 and TH1 cells; suppression and may be an effective strategy to permit more rapid and
of allergen-specific IgE and induction of IgG4 and/or higher doses of allergen immunotherapy.34 Its function on
IgA; suppression of mast cells, basophils, and eosinophils; long-term efficacy is still under investigation.
and interaction with resident tissue cells and remodeling.13 The noninflammatory role for IgG4 may arise because
the IgG4 hinge region has unique structural features that
Modulation of allergen-specific IgE and IgG result in a lower affinity for certain Fcg receptors and
subtype responses during allergen-SIT the ability to separate and repair, leading to bispecific an-
Specific IgE in serum and on the surface of mast cells tibodies that are functionally monomeric.35 Furthermore,
and basophils bound to FceRI in patients with allergy is IgG4 does not fix complement and is capable of inhibiting
a hallmark of atopic disease. Although peripheral T-cell immune-complex formation by other isotypes, giving this
tolerance is rapidly induced during SIT, there is no isotype anti-inflammatory characteristics. By using well
evidence for B-cell tolerance in the early course.10 Natural defined recombinant allergen mixtures, all treated subjects
exposure to a relevant allergen is often associated with an developed very strong allergen-specific IgG4 and also in-
increase in the IgE synthesis. Similarly, SIT frequently in- creased IgG1 antibody responses. Some patients who were
duces a transient increase in serum specific IgE, followed not initially sensitized to Phl p 5 developed strong specific
by a gradual decrease over a period of months or years of IgG4, but not IgE antibody responses to that allergen.30
treatment (Fig 1).25,26 In pollen-sensitive patients, desen- This demonstrates that extract-based antibody measure-
sitization prevents elevation of the serum specific IgE ments may provide incorrect information, and studies on
during the pollen season.27 However, the changes in IgE mechanisms of allergen-SIT should be performed with
levels cannot explain the diminished responsiveness to single allergens. Nevertheless, IgG4 antibodies can be
specific allergen as a result of SIT, because the decrease viewed as a marker of introduced allergen dose, and
in serum IgE is relatively late and does not correlate they have the ability to modulate the immune response
with clinical improvement after SIT. to allergen and thus the potential to influence the clinical
Antibody responses induced during allergen-SIT are response to allergen. In addition, the affinity of newly
functionally heterogeneous, which may account for the produced IgG4 and decreasing IgE to allergens has not
conflicting data in relation to the protective effects of been intensely studied and may have a very decisive
IgG.12,25,26 Subclasses of IgG antibodies, especially IgG4, role. Affinity maturation of specific antibodies in allergen
are thought to capture the allergen before reaching the immunotherapy and preseasonal versus postseasonal changes
effector cell-bound IgE, and thus to prevent the activation in their affinity remain to be elucidated.36
of mast cells and basophils. However, the relationship IL-10 that is induced and increasingly secreted by SIT
between the efficacy of SIT and the induction of aller- appears to counterregulate antigen-specific IgE and IgG4
gen-specific IgG subgroups remains a controversial issue, antibody synthesis.11 IL-10 is a potent suppressor of
with serum concentrations of allergen-specific IgG corre- both total and allergen-specific IgE, and it simultaneously
lating with clinical improvement in some studies but not in increases IgG4 production. Thus, IL-10 not only generates
others.28,29 Allergen-specific IgG may be directed against tolerance in T cells but also regulates specific isotype
the same epitopes as IgE, resulting in direct competition formation and skews the specific response from an IgE-
for allergen binding and a blocking effect. By contrast, dominated to an IgG4-dominated phenotype (Fig 2).
induction of IgG specific for other epitopes may result in The healthy immune response to Der p 1 demonstrated in-
a failure of the IgG response to compete with IgE, even creased specific IgA and IgG4, small amounts of IgG1, and
when IgG is present in molar excess. The concept of almost undetectable IgE antibodies in serum.12 House dust
blocking antibodies has recently been revaluated. Analysis mite–SIT did not significantly change specific IgE levels
of the IgG subtypes induced by SIT has shown specific after 70 days of treatment; however, a significant increase
increases in IgG1 and particularly IgG4, with levels in- in specific IgA, IgG1, and IgG4 was observed.12 The in-
creasing 10-fold to 100-fold.30,31 There is accumulating crease of specific IgA and IgG4 in serum coincides with in-
evidence that SIT also influences the blocking activity creased TGF-b and IL-10, respectively. This may account
on IgE-mediated responses by IgG4, and cellular as- for the role of IgA and TGF-b as well as IgG4 and IL-10
says are commonly used to investigate these changes. in peripheral mucosal immune responses to allergens in
J ALLERGY CLIN IMMUNOL Akdis and Akdis 785
VOLUME 119, NUMBER 4

feature articles
Reviews and
healthy individuals.11,37 Most probably the decrease in desensitization.47 Although its safety and efficacy have
IgE/IgG4 ratio during allergen-SIT is a feature of skew now been largely documented, much remains to be in-
from allergen-specific TH2 to Treg cell predominance. vestigated on the immunologic mechanisms underlying
However, although Treg cell generation happens within sublingual immunotherapy (SLIT). A meta-analysis of
days, a significant decrease in IgE/IgG4 ratio occurs after the double-blind, placebo-controlled trials performed in
years. The reason for the long period between the change the past decade has shown that SLIT is clinically effica-
in T-cell subsets but not IgE/IgG4 levels is not easily cious although, at present, the treatment benefit is approx-
explainable by the half-life of antibodies. In this context, imately half that achieved with subcutaneous SIT.48 The
the role of bone marrow-residing IgE-producing plasma immunologic mechanisms of sublingual SIT seem to be
cells with very long life spans remains to be investigated.38 similar to subcutaneous SIT, although the magnitude of
the change in most parameters is modest or no change
Suppression of effector cells and has been observed. It seems likely that the contact of the
inflammatory responses during allergen-SIT allergen with the oral mucosa is critical for the success of
Peripheral T-cell tolerance to allergens, which is char- SLIT.49 It is postulated that most likely oral Langerhans
acterized by functional inactivation of the cell to antigen cells are critically involved in this process.50 During
encounter, can overcome both acute and chronic events in SLIT, the allergen is captured within the oral mucosa by
allergic reactions (Fig 2). Allergen-SIT efficiently modu- oral Langerhans cells, and subsequently these cells mature
lates the thresholds for mast cell and basophil activation and migrate to proximal draining lymph nodes. Those
and decreases IgE-mediated histamine release.39 In addi- local lymph nodes may favor the production of blocking
tion, IL-10 was shown to reduce proinflammatory cytokine IgG antibodies and the induction of T lymphocytes with
release from mast cells.40 Furthermore, IL-10 downregu- suppressive function.51,52 Most studies using SLIT have
lates eosinophil function and activity and suppresses reported increased levels of serum IgG4 with a relative-
IL-5 production by human resting TH0 and TH2 cells.41 ly modest increase compared with injection immunother-
Moreover, although demonstrated in a model of myocardi- apy.53 There is no consistency in T-cell and IgE and
tis, IL-10 gene transfer significantly reduces mast cell den- effector cell responses, and a significant number of studies
sity, local histamine concentration, and mast cell growth, has failed to detect systemic immunologic changes.48,53
and prevents mast cell degranulation.42 This may be related to the different doses of allergen ad-
Long-term SIT is associated with reduction of not only ministered in different studies, or to the development of
the immediate response to allergen provocation but also more localized immunologic changes. In a recent study,
the late-phase reaction (LPR) in the nasal and bronchial PBMCs were stimulated with pollen allergen extract after
mucosa or in the skin. The mechanism of LPR is different 1 and 2 years of SLIT. The expression of IL-10 mRNA
from the mast cell–mediated immediate reaction and was increased in both high and low doses and showed a
involves the recruitment, activation, and persistence of positive correlation with TGF-b expression. IL-5 was sup-
eosinophils and activated T cells at the sites of allergen pressed with a high dose, which negatively correlated with
exposure.3 Successful SIT results not only in the increase TGF-b.54 In a recently reported birch pollen extract SLIT,
of allergen concentration necessary to induce immediate patients showed improved nasal provocation scores to
or LPR in the target tissue but also in the decreased re- birch pollen; however, apple-induced oral allergy syn-
sponses to nonspecific stimulation. Bronchial, nasal, and drome caused by cross-reactivity was not significantly re-
conjunctival hyperreactivity to nonspecific stimuli, which duced. Bet v 1–specific T-cell tolerance to all epitopes and
seems to reflect underlying mucosal inflammation, de- those cross-reactive with Mal d 1 from apple were shown.
creases after SIT and correlates with clinical improvement.43 However, neither Mal d 1–specific IgE and IgG4 levels nor
During birch pollen SIT, reduced plasma levels of eosino- Mal d 1–induced T-cell proliferation changed signifi-
phil cationic protein, a marker of eosinophil activation, as cantly, probably because of noncross-reactive epitopes.55
well as chemotactic factors for eosinophils and neutrophils These results may explain why pollen-associated food al-
correlated with decreased bronchial hyperreactivity and lergy is frequently not ameliorated by pollen immunother-
clinical improvement.44 Inhibition by SIT of the seasonal apy. Although SLIT is increasingly being used, several
increase in eosinophil priming has also been demon- points need further investigation, such as its efficacy in
strated.45 In biopsies taken during grass pollen SIT, de- asthma, its mechanisms of action, the optimal dose and
creased eosinophil and mast cell infiltration in nasal and time to be administered, its combination with injection im-
bronchial mucosa after SIT correlated with the anti-inflam- munotherapy, age of onset for its safe use in young chil-
matory effect.46 dren, and its preventive role in the development of allergy.

MECHANISMS OF SUBLINGUAL UNDERLYING MECHANISMS OF NOVEL


IMMUNOTHERAPY AND EMERGING SIT VACCINES

Because of potentially severe, albeit infrequent, side Intensive studies are being performed to improve effi-
effects associated with injection SIT, mucosal routes of cacy and safety of allergen-SIT (Table II). A basic require-
administration are being investigated to conduct allergenic ment for an allergen vaccine in achieving successful SIT
786 Akdis and Akdis J ALLERGY CLIN IMMUNOL
APRIL 2007
feature articles
Reviews and

TABLE II. Novel approaches for future of allergen-SIT

Type of the vaccine/approach Description and mechanism


59
Fusion of major allergens Several major allergens (Api m 1, Api m 2) are fused and expressed as a
single recombinant protein. IgE binding is attenuated. T-cell reactivity is
preserved. Preventive effect on generation of IgE is demonstrated in mice.
Chimeric allergens60 Fragments of major allergens (Api m 1, Api m 2, Api m 3) are fused
and expressed as a single protein. IgE binding is attenuated. T-cell
reactivity is preserved. Preventive effect on generation of IgE is
demonstrated in mice.
Fragments61 Major allergen (Bet v 1) is divided into non-IgE binding fragments. IgE
binding is attenuated. T-cell reactivity is preserved. A multicenter
clinical trial has been reported.
Peptides of major allergens58 Non-IgE binding T-cell epitope peptides (Fel d 1, Api m 1) have been
used in cat and bee venom allergy.
Polymers of major allergens61 Major allergen (Bet v 1) is trimerized. Mast cell, basophil degranulation
is attenuated. T-cell reactivity is preserved. A multi-center clinical trial
has been reported.
Unrefolded native allergens57 Major recombinant allergens (Api m 1, Bet v 1) are not refolded to the
native conformation. This decreases or abolishes IgE binding but
preserves T-cell reactivity.
Mixture of several major recombinant allergens30 Clinical efficacy of a mixture of 5 recombinant grass pollen allergens
(Phl p 1, Phl p 2, Phl p 5a, Phl p 5b, Phl p 6) in reducing symptoms
and need for symptomatic medication in patients with grass pollen
allergy was demonstrated.
GpG oligonucleotide-conjugated allergens64 Major allergen (Amb a 1) is bound to a TLR9-triggering CpG
oligonucleotide. In this way, allergen and innate immune response
stimulating agent are expressed in 1 molecule.
Allergens coupled to viruslike particles65 Der p 1 peptides coupled to highly repetitive virus capsid-like
recombinant particles induced high specific IgG titers.
Combination of conventional SIT with anti-IgE34 Anti-IgE mAb pretreatment enhances the safety of SIT for allergic
rhinitis. Its role on long-term efficacy is still under investigation.
Intralymphatic vaccination62 Allergen-SIT vaccines administered directly into a lymph node are
currently being investigated. The aim is to deliver high amounts of
allergens into secondary lymphatic organs.

without risk of anaphylaxis is to express T-cell epitopes, production have been demonstrated both in cat Fel d
which induce T-cell tolerance and lack antibody-binding 1 and bee venom Api m 1 PITs.58 A potential barrier to
sites that mediate IgE cross-linking.56 Conformation de- PIT of allergy is the apparent complexity of the aller-
pendence of B-cell epitopes and linearity of T-cell epitopes gen-specific T-cell response in terms of epitope use and
may induce a different regulation of allergen-specific dominant epitopes in human beings and stability of pep-
T-cell cytokine toward a nonallergic phenotype. Native tides. To overcome these problems, genetically engi-
allergens use IgE-facilitated antigen presentation by den- neered recombinant hybrid molecules that span the
dritic cells and B cells, which activates T cells to produce whole T-cell repertoire but do not bind IgE have been
TH2-type cytokines and B cells to produce further IgE in a developed. In cysteine-containing proteins, folding is
secondary response. In contrast, B-cell epitope deleted al- complicated by the formation of intramolecular and inter-
lergens, which do not bind IgE, do not initiate effector cell molecular disulphide bond formation, whereas any
degranulation. They use phagocytotic or pinocytic antigen formed disulphide bond can fix the conformation and
uptake mechanisms in dendritic cells, macrophages, and B limits the freedom of further folding processes. With an
cells.57 T cells may be subsequently induced to generate a increasing number of cysteines, the probability of a cor-
balanced TH0/TH1-type cytokine pattern in lower quanti- rect or nativelike folding rapidly decreases because of
ties as well as T-cell tolerance, which involves Treg cells. the increasing probability of incorrect disulfide-bound
Accordingly, targeting T cells and bypassing IgE by modi- formation. A fusion protein consisting of the 2 major al-
fied allergens will enable the administration of higher lergens of bee venom, Api m 1 and Api m 2, has been
doses of allergens, which is required to induce T-cell generated to investigate this concept. Destroyed confor-
tolerance without the risk of anaphylaxis.57 mational B-cell epitopes but intact T-cell epitopes of the
In line with this concept, immunotherapy using pep- 2 allergens characterize this protein. By providing
tides (PIT) is an attractive approach for safe SIT. To decreased allergenicity with preserved T-cell tolerance–
date, clinical trials of PIT have been performed in 2 aller- inducing capacity, the Api m [1/2] fusion protein repre-
gies.58 Induction of T-cell tolerance and increased IL-10 sents a novel vaccine prototype for allergen-SIT.59
J ALLERGY CLIN IMMUNOL Akdis and Akdis 787
VOLUME 119, NUMBER 4

feature articles
Reviews and
Another interesting approach was to cut the major aller- seasonal increase in Amb a 1–specific IgE did not occur,
gens to fragments and fuse them in a different order with- and a reduction in the number of IL-4–positive basophils
out missing any T-cell epitopes in 1 reassembled mosaic was reported.64 In another study, vaccination with a pep-
allergen.60 In this study, 2 fragments of Api m 1, 3 frag- tide antigen covalently coupled to highly repetitive virus-
ments of Api m 2, and Api m 3 are reassembled in a like particles induced high IgG antibody titers in human
different order with overlapping residues, so no T-cell beings, suggesting the possibility of using allergens cou-
epitopes are missed. Single injection of both vaccines, pled to viruslike particles in allergen-SIT.65 As a different
which only target T cells, demonstrated a preventive ef- immunologic approach, the fusion of allergens with hu-
fect on IgE generation in mice. The advantage of these man Fcg has been reported to inhibit allergen-induced
2 approaches is that only 1 molecule has to be produced basophil and mast cell degranulation by cross-linking
and purified instead of several recombinant allergens. Fcg and FceRI receptors.66
T-cell epitopes are preserved, and B-cell epitopes can be
deleted or preserved depending on the type of the fusion
molecule. Another interesting approach was to use frag- CONCLUSION
ments and a trimer of major birch pollen allergen Bet v
1 to treat birch pollen allergy. A double-blind, placebo- There is increasing evidence to support IL-10 and/or
controlled study has been completed in 3 centers, which TGF-b secreting Treg cells and immunosuppressive cy-
demonstrated an increase in IgG1, IgG2, IgG4, and IgA tokines as key players in mediating successful allergen-
and suppression of seasonal increases of IgE.61 In a differ- SIT and a healthy immune response to allergens. In
ent approach, the effectiveness of a mixture of 5 recombi- addition to allergy, these mechanisms may have implica-
nant grass pollen allergens in reducing symptoms and a tions in autoimmunity, graft-versus-host disease, tumor
need for symptomatic medication in patients with grass cell growth, parasite survival, chronic infections, and the
pollen allergy were demonstrated. All treated subjects de- development of AIDS. In sensitized individuals, periph-
veloped strong allergen-specific IgG1 and IgG4 antibody eral T-cell tolerance represents the key mechanism in
responses.30 Allergen-SIT vaccines are generally admin- healthy immune responses to allergens. Peripheral toler-
istered subcutaneously, intradermally, or sublingually, ance to allergens induced by allergen-SIT involves control
from where they must reach secondary lymphatic organs of the allergen-specific immune response in multiple
to induce an immune response. In a mouse study, an phases including specific T-cell suppression, generation of
MHC class I–binding peptide from lymphocytic chorio- noninflammatory antibody isotypes such as IgG4 and IgA,
meningitis virus enhanced immunogenicity by as much suppression of IgE and type 1 hypersensitivity responses,
as 106 times compared with subcutaneous and intradermal and suppression of LPRs and mast cells, eosinophils, and
vaccination. Intralymphatic administration induced CD8 basophils. Taking the recent advances in knowledge of
T-cell responses with strong cytotoxic activity and IFN-g peripheral tolerance mechanisms into account, develop-
production that conferred long-term protection against vi- ments of safer approaches and more efficient methods of
ral infections and tumors.62 The efficacy of allergen-SIT allergen-SIT await.
vaccines administered directly into inguinal lymph nodes New approaches to allergen-SIT, such as the use of
of human beings is currently being investigated. recombinant proteins, peptides, fragments, and hybrid
Allergen-SIT uses aluminium hydroxide as an adju- allergens, are promising, but are only in an early stage of
vant. These preparations have generally proven to be human clinical trials. There are several essential require-
efficacious and have a good safety profile, but might be ments underlying novel strategies for the development
improved in efficacy. A new class of adjuvants—so-called of safe and more efficient allergen-SIT vaccines: (1) the
immune response modifiers— act on antigen-presenting vaccine should be constituted of perfectly well standard-
cells through the Toll-like receptors (TLRs), which recog- ized native proteins or recombinant allergens; (2) the
nize pathogen-associated molecular patterns on microor- vaccine should induce tolerance in allergen-specific ef-
ganisms. Depending on the type of TLR, different types fector T cells, suppress IgE production, and promote IgG4
of antigen-presenting cells can be targeted. TLR-trigger- or IgA isotype blocking antibody production; (3) the vac-
ing compounds that can control the overexpression of cine should not induce severe side effects and should be
TH2 cytokines or skew the TH1:TH2 balance toward well tolerated; (4) the vaccine should be easily adminis-
a TH1 profile have been effective in murine models of tered; (5) the treatment should achieve clinical success
allergy.56 Oligodeoxynucleotides containing immuno- and long-term protection in a short time with few doses;
stimulatory CpG motifs that trigger TLR9, linked to the and (6) early biological/immunologic markers to assess
allergen in ragweed allergy in human beings, have been clinical success, before the onset or early in the course
used. Amb a 1–immunostimulatory DNA sequence con- of the treatment, should be identified.
jugate SIT led to a prolonged shift from TH2 immunity
toward TH1 immunity and appeared to be safe.63 In a re-
REFERENCES
cent study, the same Amb a 1 CpG conjugate was shown
1. Akdis CA. Allergy and hypersensitivity mechanisms of allergic disease.
to be effective for the treatment of allergic rhinitis in 2 con- Curr Opin Immunol 2006;18:718-26.
secutive seasons. Although an early increase in Amb a 1– 2. Romagnani S. Immunologic influences on allergy and the TH1/TH2
specific IgE was observed during the injection phase, a balance. J Allergy Clin Immunol 2004;113:395-400.
788 Akdis and Akdis J ALLERGY CLIN IMMUNOL
APRIL 2007
feature articles
Reviews and

3. Larche M, Akdis CA, Valenta R. Immunological mechanisms of aller- grass pollen allergens (Lol p 1, 2, 3 and 5) during immunotherapy.
gen-specific immunotherapy. Nat Rev Immunol 2006;6:761-71. Clin Exp Allergy 1997;27:68-74.
4. Akdis CA, Blaser K, Akdis M. Apoptosis in tissue inflammation and 26. Gleich GJ, Zimmermann EM, Henderson LL, Yunginger JW. Effect of
allergic disease. Curr Opin Immunol 2004;16:717-23. immunotherapy on immunoglobulin E and immunoglobulin G antibodies
5. Trautmann A, Schmid-Grendelmeier P, Krüger K, Crameri R, Akdis M, to ragweed antigens: a six-year prospective study. J Allergy Clin Immu-
Akkaya A, et al. T cells and eosinophils cooperate in the induction of nol 1982;70:261-71.
bronchial epithelial apoptosis in asthma. J Allergy Clin Immunol 2002; 27. Bousquet J, Maasch H, Martinot B, Hejjaoui A, Wahl R, Michel FB.
109:329-37. Double-blind, placebo-controlled immunotherapy with mixed grass-pol-
6. Woo HY, Kim YS, Kang NI, Chung WC, Song CH, Choi IW, et al. len allergoids, II: comparison between parameters assessing the efficacy
Mechanism for acute oral desensitization to antibiotics. Allergy 2006; of immunotherapy. J Allergy Clin Immunol 1988;82:439-46.
61:954-8. 28. Golden DB, Meyers DA, Kagey-Sobotka A, Valentine MD, Lichtenstein
7. Eberlein-Konig B, Ullmann S, Thomas P, Przybilla B. Tryptase and his- LM. Clinical relevance of the venom-specific immunoglobulin G anti-
tamine release due to a sting challenge in bee venom allergic patients body level during immunotherapy. J Allergy Clin Immunol 1982;69:
treated successfully or unsuccessfully with hyposensitization. Clin Exp
489-93.
Allergy 1995;25:704-12.
29. Müller UR, Helbling A, Bischof M. Predictive value of venom-specific
8. Jutel M, Muller UR, Fricker M, Rihs S, Pichler WJ, Dahinden C. Influ-
IgE, IgG and IgG subclass antibodies in patients on immunotherapy
ence of bee venom immunotherapy on degranulation and leukotriene
with honey bee venom. Allergy 1989;44:412-8.
generation in human blood basophils. Clin Exp Allergy 1996;26:1112-8.
30. Jutel M, Jaeger L, Suck R, Meyer H, Fiebig H, Cromwell O. Allergen-
9. Plewako H, Wosinska K, Arvidsson M, Bjorkander J, Skov PS, Hakans-
specific immunotherapy with recombinant grass pollen allergens. J Al-
son L, et al. Basophil interleukin 4 and interleukin 13 production is sup-
lergy Clin Immunol 2005;116:608-13.
pressed during the early phase of rush immunotherapy. Int Arch Allergy
31. Reisinger J, Horak F, Pauli G, van Hage M, Cromwell O, Konig F, et al.
Immunol 2006;141:346-53.
10. Akdis CA, Akdis M, Blesken T, Wymann D, Alkan SS, Müller U, et al. Allergen-specific nasal IgG antibodies induced by vaccination with ge-
Epitope specific T cell tolerance to phospholipase A2 in bee venom im- netically modified allergens are associated with reduced nasal allergen
munotherapy and recovery by IL-2 and IL-15 in vitro. J Clin Invest 1996; sensitivity. J Allergy Clin Immunol 2005;116:347-54.
98:1676-83. 32. Wachholz PA, Durham SR. Mechanisms of immunotherapy: IgG revis-
11. Akdis CA, Blesken T, Akdis M, Wüthrich B, Blaser K. Role of IL-10 in ited. Curr Opin Allergy Clin Immunol 2004;4:313-8.
specific immunotherapy. J Clin Invest 1998;102:98-106. 33. Till SJ, Francis JN, Nouri-Aria K, Durham SR. Mechanisms of immuno-
12. Jutel M, Akdis M, Budak F, Aebischer-Casaulta C, Wrzyszcz M, Blaser therapy. J Allergy Clin Immunol 2004;113:1025-34; quiz 35.
K, et al. IL-10 and TGF-b cooperate in regulatory T cell response to 34. Casale TB, Busse WW, Kline JN, Ballas ZK, Moss MH, Townley RG,
mucosal allergens in normal immunity and specific immunotherapy. et al. Omalizumab pretreatment decreases acute reactions after rush
Eur J Immunol 2003;33:1205-14. immunotherapy for ragweed-induced seasonal allergic rhinitis. J Allergy
13. Akdis M, Blaser K, Akdis CA. T regulatory cells in allergy: novel con- Clin Immunol 2006;117:134-40.
cepts in the pathogenesis, prevention, and treatment of allergic diseases. 35. Aalberse RC, Schuurman J. IgG4 breaking the rules. Immunology 2002;
J Allergy Clin Immunol 2005;116:961-8; quiz 9. 105:9-19.
14. Jutel M, Akdis M, Budak F, Aebischer-Casaulta C, Wrzyszcz M, Blaser 36. Poirier MP, Ahlstedt S, Ford J, Dolen WK. Use of thiocyanate elution
K, et al. IL-10 and TGF-beta cooperate in the regulatory T cell response to estimate relative avidity of allergen specific IgE antibodies. Allergy
to mucosal allergens in normal immunity and specific immunotherapy. Asthma Proc 1997;18:359-62.
Eur J Immunol 2003;33:1205-14. 37. Sonoda E, Matsumoto R, Hitoshi Y, Ishii T, Sugimoto M, Araki S, et al.
15. Francis JN, Till SJ, Durham SR. Induction of IL-101CD41CD251 T Transforming growth factor beta induces IgA production and acts addi-
cells by grass pollen immunotherapy. J Allergy Clin Immunol 2003; tively with interleukin 5 for IgA production. J Exp Med 1989;170:
111:1255-61. 1415-20.
16. Ling EM, Smith T, Nguyen XD, Pridgeon C, Dallman M, Arbery J, et al. 38. Radbruch A, Muehlinghaus G, Luger EO, Inamine A, Smith KG, Dorner
Relation of CD41CD251 regulatory T-cell suppression of allergen- T, et al. Competence and competition: the challenge of becoming a long-
driven T-cell activation to atopic status and expression of allergic disease. lived plasma cell. Nat Rev Immunol 2006;6:741-50.
Lancet 2004;363:608-15. 39. Pierkes M, Bellinghausen I, Hultsch T, Metz G, Knop J, Saloga J. De-
17. Schmidt-Weber CB, Blaser K. Regulation and role of transforming creased release of histamine and sulfidoleukotrienes by human peripheral
growth factor-beta in immune tolerance induction and inflammation. blood leukocytes after wasp venom immunotherapy is partially due to
Curr Opin Immunol 2004;16:709-16. induction of IL-10 and IFN-gamma production of T cells. J Allergy
18. Akdis M, Verhagen J, Taylor A, Karamloo F, Karagiannidis C, Crameri
Clin Immunol 1999;103:326-32.
R, et al. Immune responses in healthy and allergic individuals are char-
40. Marshall JS, Leal-Berumen I, Nielsen L, Glibetic M, Jordana M. Interleu-
acterized by a fine balance between allergen-specific T regulatory 1 and
kin (IL)-10 Inhibits long-term IL-6 production but not preformed mediator
T helper 2 cells. J Exp Med 2004;199:1567-75.
release from rat peritoneal mast cells. J Clin Invest 1996;97:1122-8.
19. Akdis M. Healthy immune response to allergens: T regulatory cells and
41. Schandane L, Alonso-Vega C, Willems F, Gerard C, Delvaux A, Velu T,
more. Curr Opin Immunol 2006;18:738-44.
et al. B7/CD28-dependent IL-5 production by human resting T cells is
20. Karlsson MR, Rugtveit J, Brandtzaeg P. Allergen-responsive CD41CD251
inhibited by IL-10. J Immunol 1994;152:4368-74.
regulatory T cells in children who have outgrown cow’s milk allergy. J Exp
42. Palaniyandi SS, Watanabe K, Ma M, Tachikawa H, Kodama M, Aizawa Y.
Med 2004;199:1679-88.
21. Cavani A, Nasorri F, Ottaviani C, Sebastiani S, De Pita O, Girolomoni G. Inhibition of mast cells by interleukin-10 gene transfer contributes to pro-
Human CD251 regulatory T cells maintain immune tolerance to nickel tection against acute myocarditis in rats. Eur J Immunol 2004;34:3508-15.
in healthy, nonallergic individuals. J Immunol 2003;171:5760-8. 43. Rak S, Lowhagen O, Venge P. The effect of immunotherapy on bron-
22. Prakken BJ, Samodal R, Le TD, Giannoni F, Yung GP, Scavulli J, et al. chial hyperresponsiveness and eosinophil cationic protein in pollen-
Epitope-specific immunotherapy induces immune deviation of proinflam- allergic patients. J Allergy Clin Immunol 1988;82:470-80.
matory T cells in rheumatoid arthritis. Proc Natl Acad Sci U S A 2004; 44. Rak S, Hakanson L, Venge P. Immunotherapy abrogates the generation
101:4228-33. of eosinophil and neutrophil chemotactic activity during pollen season.
23. Chatila TA. Role of regulatory T cells in human diseases. J Allergy Clin J Allergy Clin Immunol 1990;86:706-13.
Immunol 2005;116:949-59; quiz 60. 45. Hakansson L, Heinrich C, Rak S, Venge P. Priming of eosinophil adhe-
24. Verhagen J, Akdis M, Traidl-Hoffmann C, Schmid-Grendelmeier P, Hij- sion in patients with birch pollen allergy during pollen season: effect of
nen D, Knol EF, et al. Absence of T-regulatory cell expression and func- immunotherapy. J Allergy Clin Immunol 1997;99:551-62.
tion in atopic dermatitis skin. J Allergy Clin Immunol 2006;117:176-83. 46. Creticos PS, Adkinson NF Jr, Kagey-Sobotka A, Proud D, Meier HL,
25. Van Ree R, Van Leeuwen WA, Dieges PH, Van Wijk RG, De Jong N, Naclerio RM, et al. Nasal challenge with ragweed pollen in hay fever
Brewczyski PZ, et al. Measurement of IgE antibodies against purified patients: effect of immunotherapy. J Clin Invest 1985;76:2247-53.
J ALLERGY CLIN IMMUNOL Akdis and Akdis 789
VOLUME 119, NUMBER 4

feature articles
Reviews and
47. Passalacqua G, Guerra L, Compalati E, Fumagalli F, Cirillo A, Canonica 57. Akdis CA, Blaser K. Bypassing IgE and targeting T cells for specific
GW. New insights in sublingual immunotherapy. Curr Allergy Asthma immunotherapy of allergy. Trends Immunol 2001;22:175-8.
Rep 2006;6:407-12. 58. Larche M. Immunoregulation by targeting T cells in the treatment of
48. Wilson DR, Lima MT, Durham SR. Sublingual immunotherapy for aller- allergy and asthma. Curr Opin Immunol 2006;18:745-50.
gic rhinitis: systematic review and meta-analysis. Allergy 2005;60:4-12. 59. Kussebi F, Karamloo F, Rhyner C, Schmid-Grendelmeier P, Salagianni
49. Passalacqua G, Albano M, Fregonese L, Riccio A, Pronzato C, Mela GS, M, Mannhart C, et al. A major allergen gene-fusion protein for potential
et al. Randomized controlled trial of local allergoid immunotherapy on usage in allergen-specific immunotherapy. J Allergy Clin Immunol 2005;
allergic inflammation in mite-induced rhinoconjunctivitis. Lancet 1998; 115:323-9.
351:629-32. 60. Karamloo F, Schmid-Grendelmeier P, Kussebi F, Akdis M, Salagianni
50. Allam JP, Niederhagen B, Bucheler M, Appel T, Betten H, Bieber T, M, von Beust BR, et al. Prevention of allergy by a recombinant multi-
et al. Comparative analysis of nasal and oral mucosa dendritic cells. allergen vaccine with reduced IgE binding and preserved T cell epitopes.
Allergy 2006;61:166-72. Eur J Immunol 2005;35:3268-76.
51. Samsom JN, van Berkel LA, van Helvoort JM, Unger WW, Jansen W, 61. Niederberger V, Horak F, Vrtala S, Spitzauer S, Krauth MT, Valent P,
et al. Vaccination with genetically engineered allergens prevents progres-
Thepen T, et al. Fc gamma RIIB regulates nasal and oral tolerance: a
sion of allergic disease. Proc Natl Acad Sci U S A 2004;101(suppl 2):
role for dendritic cells. J Immunol 2005;174:5279-87.
14677-82.
52. van Helvoort JM, Samsom JN, Chantry D, Jansen W, Schadee-Eester-
62. Johansen P, Haffner AC, Koch F, Zepter K, Erdmann I, Maloy K, et al.
mans I, Thepen T, et al. Preferential expression of IgG2b in nose draining
Direct intralymphatic injection of peptide vaccines enhances immunoge-
cervical lymph nodes and its putative role in mucosal tolerance induction.
nicity. Eur J Immunol 2005;35:568-74.
Allergy 2004;59:1211-8.
63. Simons FE, Shikishima Y, Van Nest G, Eiden JJ, HayGlass KT. Selec-
53. Moingeon P, Batard T, Fadel R, Frati F, Sieber J, Van Overtvelt L.
tive immune redirection in humans with ragweed allergy by injecting
Immune mechanisms of allergen-specific sublingual immunotherapy. Amb a 1 linked to immunostimulatory DNA. J Allergy Clin Immunol
Allergy 2006;61:151-65. 2004;113:1144-51.
54. Savolainen J, Jacobsen L, Valovirta E. Sublingual immunotherapy in 64. Creticos PS, Schroeder JT, Hamilton RG, Balcer-Whaley SL, Khattigna-
children modulates allergen-induced in vitro expression of cytokine vong AP, Lindblad R, et al. Immunotherapy with a ragweed-toll-like re-
mRNA in PBMC. Allergy 2006;61:1184-90. ceptor 9 agonist vaccine for allergic rhinitis. N Engl J Med 2006;355:
55. Kinaciyan T, Jahn-Schmid B, Radakovics A, Zwolfer B, Schreiber C, 1445-55.
Francis JN, et al. Successful sublingual immunotherapy with birch pollen 65. Kundig TM, Senti G, Schnetzler G, Wolf C, Prinz Vavricka BM, Fulurija
has limited effects on concomitant food allergy to apple and the immune A, et al. Der p 1 peptide on virus-like particles is safe and highly immu-
response to the Bet v 1 homolog Mal d 1. J Allergy Clin Immunol 2007; nogenic in healthy adults. J Allergy Clin Immunol 2006;117:1470-6.
Jan 3 [epub ahead of print]. 66. Zhu D, Kepley CL, Zhang M, Zhang K, Saxon A. A novel human immu-
56. Crameri R, Rhyner C. Novel vaccines and adjuvants for allergen-specific noglobulin Fc gamma Fc epsilon bifunctional fusion protein inhibits
immunotherapy. Curr Opin Immunol 2006;18:761-8. Fc epsilon RI-mediated degranulation. Nat Med 2002;8:518-21.

Vous aimerez peut-être aussi